CN113242858A - Preparation and application of protein receptor kinase inhibitor - Google Patents

Preparation and application of protein receptor kinase inhibitor Download PDF

Info

Publication number
CN113242858A
CN113242858A CN201980067895.1A CN201980067895A CN113242858A CN 113242858 A CN113242858 A CN 113242858A CN 201980067895 A CN201980067895 A CN 201980067895A CN 113242858 A CN113242858 A CN 113242858A
Authority
CN
China
Prior art keywords
substituted
unsubstituted
ring
compound
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201980067895.1A
Other languages
Chinese (zh)
Inventor
江磊
冯志勇
路小丽
尚珂
寿建勇
吴淡宜
徐圆
许灵灵
张淑芸
张宇星
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Ennovabio Pharmaceuticals Co Ltd
Original Assignee
Shanghai Ennovabio Pharmaceuticals Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Ennovabio Pharmaceuticals Co Ltd filed Critical Shanghai Ennovabio Pharmaceuticals Co Ltd
Publication of CN113242858A publication Critical patent/CN113242858A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/527Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim spiro-condensed
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Abstract

The invention provides a preparation method of a protein receptor kinase inhibitor. Specifically, the invention discloses a compound shown as a formula I, or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof, wherein the definition of each group is described in the specification. The invention also discloses application of the compound as a TRK kinase inhibitor.

Description

Preparation and application of protein receptor kinase inhibitor Technical Field
The invention relates to the technical field of medicines, and particularly relates to a protein receptor kinase inhibitor.
Background
Tropomyosin-receptor kinase (TRK) is a nerve growth factor receptor, belongs to a receptor tyrosine kinase family, mainly comprises three highly homologous members of TRKA, TRKB and TRKC, and is respectively encoded by three genes of NTRK1, NTRK2 and NTRK 3. These receptor tyrosine kinases are mainly expressed in nervous tissues and play an important role in development and physiological functions of the nervous system through activation of neurotrophic factors nts (neurotropins). TRK function as tyrosine kinase receptors, each TRK having a signaling pathway that binds to and activates its downstream ligand. NGF (nerve growth factor) specifically binds to and activates TRKA; the TRKB ligand comprises BDGF (broad-derived growth factor) and NT-4/5 (neurotropin-4/5); NT-3 specifically binds to and activates TRKC. All three TRK receptors contain an extracellular domain for ligand binding, a transmembrane domain, and an intracellular domain with kinase activity.
When a specific ligand is combined with an extracellular domain of a corresponding receptor, oligomerization of the receptor and phosphorylation of specific tyrosine residues in an intracytoplasmic kinase domain are triggered, so that downstream signal paths such as Ras/MAPK, PLC gamma/PKC and PI3K/AKT signal paths are activated, and a series of physiological processes such as proliferation, differentiation and survival of nerve cells are regulated. The TRK signaling pathway is usually precisely regulated, and its abnormal activation is closely related to tumorigenesis. Research results show that there are many mechanisms causing abnormal activation of TRK pathway, including gene fusion, protein overexpression and single nucleotide mutation, and these abnormalities are closely related to the pathogenesis of tumor, especially NTRK gene fusion has been proved to be an important factor causing multiple tumorigenesis, and is independent of the tissue source and type of tumor. Under the rapid development of current second-generation sequencing technologies and precision medicine, more and more NTRK fusion genes are found, such as ETV6-NTRK3, MPRIP-NTRK 1, CD 74-NTRK 1 and the like. The results of recent clinical trials show that the fusion genes are very effective anti-cancer targets, and the tumors containing the NTRK fusion genes have very significant response rate to TRK inhibitors.
Therefore, more and more TRK target inhibitors are reported, e.g. (WO2010048314, WO2011146336, WO 2017004342). Meanwhile, in the clinical trial stage, it has been found that some of the patients receiving the treatment have a drug resistance phenomenon and proved to be caused by partial base mutation of the enzyme activity region, such as mutation of NTRK 1G 595R or G667C, G623R or G696A of NTRK 3. Therefore, there is still a need in the art to develop a new generation of TRK kinase inhibitors to solve the above problems.
Disclosure of Invention
The invention aims to provide a novel TRK kinase inhibitor, a preparation method and application thereof.
In a first aspect of the present invention, there is provided a compound represented by formula I, or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof:
Figure PCTCN2019111334-APPB-000001
wherein the content of the first and second substances,
ring a has a structure represented by the following formula:
Figure PCTCN2019111334-APPB-000002
each X4Each independently is C or N;
each X5Each independently is CR7Or N; wherein R is7Is hydrogen, deuterium, halogen, NH2Or OH;
R 1、R 2taken together with the nitrogen atom to which they are attached to form a 6-12 membered fused ring (including bicyclic fused ring, tricyclic fused ring), bridged ring or spiro ring (including substituted or unsubstituted fused rings (including bicyclic fused ring, tricyclic fused ring), bridged ring or spiro ring); wherein said fused, bridged or spiro ring contains 1 to 3 heteroatoms selected from nitrogen atoms, oxygen atoms and sulfur atoms;
R Ais composed of
Figure PCTCN2019111334-APPB-000003
Wherein the content of the first and second substances,
X 1、X 2、X 3each independently is CR6Or N; r6Is hydrogen, deuterium, hydroxy, halogen, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy;
n is 1,2 or 3;
each R is3Each independently is hydrogen, deuterium, hydroxy, halogen, trifluoromethyl, cyano, amino, amido, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy;
or n is 2 and two R3co-form-O- (CH)2) m-O-; m is an integer of 1 to 4;
R Bis a substituted or unsubstituted 4-7 membered heterocyclic ring (said heterocyclic ring containing 1 to 3 hetero atoms selected from the group consisting of a nitrogen atom, an oxygen atom and a sulfur atom); - (CH)2) m1-、-O-(CH 2) m2-O-; wherein m1 and m2 are each independently an integer of 1 to 4
Or RBIs-substituted or unsubstituted-C1-C10 alkylene-L2- (wherein, L)2Is NH, O, S or S (O)2) Substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy, -O- (CH)2) m-O-; m is an integer of 1 to 4;
unless otherwise specified, "substituted" means substituted with one or more (e.g., 2, 3,4, etc.) substituents selected from the group consisting of: deuterium, halogen, C1-C6 alkoxy, halogenated C1-C6 alkyl, halogenated C1-C6 alkoxy, halogenated C3-C8 cycloalkyl, methylsulfonyl, -S (═ O)2NH 2Oxo (═ O), -CN, hydroxy, -NH2Carboxy, C1-C6 amido (-C (═ O) -N (Rc)2or-NH-C (═ O) (Rc), Rc being H or C1-C5 alkyl), C1-C6 alkyl- (C1-C6 amido),
Figure PCTCN2019111334-APPB-000004
Or a substituted or unsubstituted group selected from: C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 amino, C6-C10 aryl, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O, 5-12 membered heterocyclyl having 1-3 heteroatoms selected from N, S and O, - (CH) 6 alkyl2) -C6-C10 aryl, - (CH)2) - (5-to 10-membered heteroaryl having 1 to 3 heteroatoms selected from N, S and O), and the substituents are selected from the group consisting of: halogen, C1-C6 alkyl, C1-C6 alkoxy, oxo, -CN, -NH2-OH, C6-C10 aryl, C1-C6 amine, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O;
the dotted line represents a chemical bond or nothing.
In another preferred embodiment, the compound of formula I has the structure shown in formula I-1 below:
Figure PCTCN2019111334-APPB-000005
in another preferred embodiment, ring a is a bicyclic 9-10 membered heteroaromatic ring; wherein the heteroaromatic ring contains 1-3 nitrogen atoms.
In another preferred embodiment, ring a is selected from the group consisting of:
Figure PCTCN2019111334-APPB-000006
in another preferred embodiment, the ring a is selected from the group consisting of:
Figure PCTCN2019111334-APPB-000007
in another preferred embodiment, R1、R 2Together with the nitrogen atom to which they are attached form a substituted or unsubstituted 6-12 membered bicyclic, bridged or spirocyclic ring; the ring combinationThe bridged or spiro ring contains 1-3 nitrogen atoms.
In another preferred embodiment, the fused ring is a fused ring selected from the group consisting of: 3 and 5, 4 and 5, 5 and 5, 6 and 5, 7 and 5, 6 and 6, 6 and 7.
In another preferred embodiment, the spiro ring is selected from the following group: spiro [4.5] ring, spiro [ 5.5 ] ring, spiro [ 5.6 ] ring, spiro [ 5.7 ] ring.
In another preferred embodiment, R1、R 2Together with the nitrogen atom to which they are attached form a substituted or unsubstituted group selected from:
Figure PCTCN2019111334-APPB-000008
Figure PCTCN2019111334-APPB-000009
in another preferred embodiment, said substitution means substitution by one or more groups selected from the group consisting of: C1-C6 alkyl, oxo, C1-C6 alkylcyano.
In another preferred embodiment, substituted
Figure PCTCN2019111334-APPB-000010
The method comprises the following steps:
Figure PCTCN2019111334-APPB-000011
Figure PCTCN2019111334-APPB-000012
in another preferred embodiment, substituted
Figure PCTCN2019111334-APPB-000013
Included
Figure PCTCN2019111334-APPB-000014
In another preferred embodiment, substituted
Figure PCTCN2019111334-APPB-000015
Included
Figure PCTCN2019111334-APPB-000016
In another preferred embodiment, substituted
Figure PCTCN2019111334-APPB-000017
Included
Figure PCTCN2019111334-APPB-000018
In another preferred embodiment, substituted
Figure PCTCN2019111334-APPB-000019
Included
Figure PCTCN2019111334-APPB-000020
In another preferred embodiment, substituted
Figure PCTCN2019111334-APPB-000021
Included
Figure PCTCN2019111334-APPB-000022
Figure PCTCN2019111334-APPB-000023
In another preferred embodiment, RASelected from the group consisting of:
Figure PCTCN2019111334-APPB-000024
in each formula, X1、X 2The definition is the same as the previous definition;
on the aromatic ring, n is 1,2 or 3; and R is3Is hydrogen, deuterium, hydroxy, halogen, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy; or n is 2 and two R3co-form-O- (CH)2) m-O-; m is an integer of 1 to 4;
r elsewhere3、R 4、R 5Each independently hydrogen, deuterium, hydroxy, halogen, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy.
In another preferred embodiment, RAIs composed of
Figure PCTCN2019111334-APPB-000025
Wherein, X1Is CR6Or N; x2、X 3Each independently is CR6
n is 1,2 or 3; and R is3Is hydrogen, deuterium, hydroxy, halogen, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy; (ii) a Or n is 2 and two R3co-form-O- (CH)2) m-O-; m is an integer of 1 to 4.
In another preferred embodiment, the compound of formula I is selected from the group consisting of:
Figure PCTCN2019111334-APPB-000026
Figure PCTCN2019111334-APPB-000027
Figure PCTCN2019111334-APPB-000028
in another preferred embodiment, the compound of formula I is selected from the group consisting of:
Figure PCTCN2019111334-APPB-000029
Figure PCTCN2019111334-APPB-000030
in a second aspect of the invention, there is provided a pharmaceutical composition comprising a therapeutically effective amount of a compound according to the first aspect of the invention or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
In a third aspect of the present invention, there is provided a use of the compound according to the first aspect of the present invention or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition according to the second aspect of the present invention, in a medicament for preventing and treating a TRK-associated disease.
In another preferred embodiment, the TRK-associated disease is selected from the group consisting of: cancer, proliferative diseases, pain, skin diseases or conditions, metabolic diseases, muscular diseases, neurological diseases, autoimmune diseases, dermatitis-induced pruritus, inflammation-related diseases, bone-related diseases.
In another preferred embodiment, the cancer is selected from cancers associated with abnormal TRK function (abnormal TRK activation due to amplification, overexpression, mutation, or gene fusion), including but not limited to: neuroblastoma, prostate cancer, thyroid cancer, lung cancer, ovarian cancer, pancreatic cancer, colorectal cancer, non-small cell lung cancer, fibrosarcoma, and the like.
In a fourth aspect of the present invention, there is provided a use of a compound according to the first aspect of the present invention or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, or a pharmaceutical composition according to the second aspect of the present invention, for the preparation of a pharmaceutical composition for the prevention and/or treatment of a disease associated with abnormal TRK function (abnormal TRK activation due to amplification, or overexpression, or mutation, or gene fusion).
In another preferred embodiment, the pharmaceutical composition may be administered in combination with other therapeutic agents.
In another preferred embodiment, the drug combination includes, but is not limited to, other target kinase inhibitors, immunomodulators (tumor immune checkpoint inhibitors), cell division blockers and other conventional first-line chemotherapeutic drugs.
In another preferred embodiment, the disease is selected from the group consisting of: cancer, proliferative diseases, pain, skin diseases or conditions, metabolic diseases, muscular diseases, neurological diseases, autoimmune diseases, dermatitis-induced pruritus.
In another preferred embodiment, the cancer is selected from cancers associated with abnormal TRK function (abnormal TRK activation due to amplification, overexpression, mutation, or gene fusion), including but not limited to: neuroblastoma, prostate cancer, thyroid cancer, lung cancer, ovarian cancer, pancreatic cancer, colorectal cancer, non-small cell lung cancer, fibrosarcoma, and the like.
In a fifth aspect of the invention, there is provided a TRK inhibitor comprising a compound according to the first aspect of the invention, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof.
In another preferred embodiment, the TRK inhibitor selectively inhibits one or more TRK kinases selected from the group consisting of: TRKA, TRKB or TRKC.
In a sixth aspect of the present invention, there is provided a process for the preparation of a compound according to the first aspect of the present invention or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
Figure PCTCN2019111334-APPB-000031
reacting a compound of formula Ia with a compound of formula Ib in an inert solvent, thereby forming a compound of formula I;
wherein R is1Is C1-C6 alkyl; the remaining groups are as defined for the first aspect of the invention.
In another preferred embodiment, when the compound of formula Ib further comprises a protecting group (e.g., an amino protecting group or a hydroxyl protecting group), this step further comprises a deprotection step.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Detailed Description
The inventor discovers a novel and effective TRK kinase small molecule inhibitor for the first time through extensive and intensive research. The present invention has been completed based on this finding.
Term(s) for
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
As used herein, the term "comprising" or "includes" can be open, semi-closed, and closed. In other words, the term also includes "consisting essentially of …," or "consisting of ….
In this application, the term "alkyl" as a group or part of another group refers to a fully saturated straight or branched hydrocarbon chain radical consisting of only carbon and hydrogen atoms, having, for example, from 1 to 12 (preferably from 1 to 8, more preferably from 1 to 6) carbon atoms, and attached to the rest of the molecule by a single bond, including, for example, but not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl, decyl, and the like. For the purposes of the present invention, the term "C1-C6 alkyl" refers to alkyl groups containing from 1 to 6 carbon atoms.
In this application, the term "6-to 10-membered aromatic ring" as a group or as part of another group means an aromatic ring having 6 to 10 ring atoms, which are carbon atoms. The aromatic ring may be monocyclic or bicyclic. Such as benzene rings, naphthalene rings, and the like.
In this application, the term "5-to 10-membered heteroaromatic ring" as a group or as part of another group means a heteroaromatic ring having 5 to 10 ring atoms, at least 1 (which may be 1,2 or 3) of which is a heteroatom selected from nitrogen, oxygen and sulfur. The heteroaryl ring may be monocyclic or bicyclic. For example, a pyrimidopyrazole ring, a pyrazinoimidazole ring, a pyridopyrazole ring, a pyridoimidazole ring, a pyridopyrimidine ring, a pyridopyridine ring.
In this application, the term "6-11 membered bridged or spiro ring" as a group or as part of another group means a bicyclic ring having 6-11 ring atoms, and wherein the two rings are bridged or spiro-linked. The bridged or spiro ring contains 1 to 3 heteroatoms selected from nitrogen atoms, oxygen atoms, and sulfur atoms. For example,
Figure PCTCN2019111334-APPB-000032
Figure PCTCN2019111334-APPB-000033
compounds of the invention
The compound is a compound shown in a formula I, or a stereoisomer or racemate thereof, or pharmaceutically acceptable salt thereof.
The compounds of the present invention may contain one or more chiral carbon atoms and may therefore give rise to enantiomers, diastereomers and other stereoisomeric forms. Each chiral carbon atom may be defined as (R) -or (S) -, based on stereochemistry. The present invention is intended to include all possible isomers, as well as racemates and optically pure forms thereof. The compounds of the invention may be prepared by selecting as starting materials or intermediates racemates, diastereomers or enantiomers. Optically active isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, e.g., crystallization and chiral chromatography.
Conventional techniques for the preparation/separation of individual isomers include Chiral synthesis from suitable optically pure precursors, or resolution of racemates (or racemates of salts or derivatives) using, for example, Chiral high performance liquid chromatography, as described, for example, in Gerald Gubitz and Martin G.Schmid (Eds.), Chiral Separations, Methods and Protocols, Methods in Molecular Biology, Vol.243, 2004; m. Stalcup, Chiral Separations, Annu. Rev. anal. chem.3:341-63, 2010; fumiss et al (eds.), VOGEL' S ENCYCOPEDIA OF PRACTICAL ORGANIC CHEMISTRY 5. TH ED., Longman Scientific and Technical Ltd., Essex,1991, 809-816; heller, acc, chem, res, 1990,23,128.
The term "pharmaceutically acceptable salts" includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
"pharmaceutically acceptable acid addition salts" refers to salts with inorganic or organic acids which retain the biological effectiveness of the free base without other side effects. Inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate, and the like; organic acid salts include, but are not limited to, formates, acetates, 2-dichloroacetates, trifluoroacetates, propionates, caproates, caprylates, caprates, undecylenates, glycolates, gluconates, lactates, sebacates, adipates, glutarates, malonates, oxalates, maleates, succinates, fumarates, tartrates, citrates, palmitates, stearates, oleates, cinnamates, laurates, malates, glutamates, pyroglutamates, aspartates, benzoates, methanesulfonates, benzenesulfonates, p-toluenesulfonates, alginates, ascorbates, salicylates, 4-aminosalicylates, napadisylates, and the like. These salts can be prepared by methods known in the art.
"pharmaceutically acceptable base addition salts" refers to salts with inorganic or organic bases which maintain the biological effectiveness of the free acid without other side effects. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, the following: primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. These salts can be prepared by methods known in the art.
Preparation method
The following reaction schemes illustrate methods for preparing compounds of formula I, or stereoisomers or racemates thereof, or pharmaceutically acceptable salts thereof, wherein each group is as described above. It is understood that in the following reaction schemes, combinations of substituents and/or variables in the general formulae are permissible only if such combinations result in stable compounds. It will also be appreciated that other general formulae may be prepared by methods disclosed herein (by applying appropriately substituted starting materials and modifying the synthesis parameters as required using methods well known to those skilled in the art) or known methods by those skilled in the art of organic chemistry.
The method comprises the following steps:
Figure PCTCN2019111334-APPB-000034
reacting a compound of formula Ia with a compound of formula Ib in an inert solvent, thereby forming a compound of formula I;
wherein R is1Is C1-C6 alkyl; ring B is as defined for the first aspect of the invention.
In another preferred embodiment, when the compound of formula Ib further comprises a protecting group (e.g., an amino protecting group or a hydroxyl protecting group), this step further comprises a deprotection step.
It will also be appreciated by those skilled in the art that in the processes described below, the functional groups of the intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxyl, amino, mercapto and carboxylic acid. Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g.tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butyloxycarbonyl, benzyloxycarbonyl and the like. Suitable thiol protecting groups include-C (O) -R "(where R" is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters.
Protecting groups may be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protecting Groups is described in detail in Greene, T.W. and P.G.M.Wuts, Protective Groups in organic Synthesis, (1999),4th Ed., Wiley. The protecting group may also be a polymeric resin.
Applications of
The compound of the present invention has excellent TRK (e.g., TRKA, TRKB, TRKC) inhibitory activity, and thus the compound of the present invention and various crystalline forms, pharmaceutically acceptable inorganic or organic salts, hydrates, or solvates thereof, and a pharmaceutical composition containing the compound of the present invention as a main active ingredient may be used for the prevention and/or treatment of diseases associated with TRK kinase activity or expression amount (e.g., cancer).
In the present application, the term "pharmaceutical composition" refers to a formulation of a compound of the present invention with a vehicle generally accepted in the art for delivery of biologically active compounds to a mammal (e.g., a human). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of active ingredients and exert biological activity.
In the present application, the term "pharmaceutically acceptable" refers to a substance (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention and is relatively non-toxic, i.e., the substance can be administered to an individual without causing an adverse biological response or interacting in an adverse manner with any of the components contained in the composition.
In this application, the term "pharmaceutically acceptable excipient" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifying agent that is approved by the relevant governmental regulatory agency for human or livestock use.
In the present application, the term "tumor" includes, but is not limited to, glioma, sarcoma, melanoma, arthroscleroma, cholangioma, leukemia, gastrointestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, pancreatic cancer, lung squamous cancer, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, nasopharyngeal cancer, brain cancer, bone cancer, esophageal cancer, melanoma, renal cancer, oral cancer, and the like.
In this application, the terms "preventing", "prevention" and "prevention" include reducing the likelihood of occurrence or worsening of a disease or disorder in a subject.
In the present application, the term "treatment" and other similar synonyms include the following meanings:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease or condition, but has not been diagnosed as having the disease or condition;
(ii) inhibiting the disease or disorder, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the state of the disease or condition; or
(iv) Alleviating the symptoms caused by the disease or disorder.
In the present application, the term "effective amount", "therapeutically effective amount" or "pharmaceutically effective amount" refers to an amount of at least one agent or compound that is sufficient to alleviate one or more symptoms of the disease or condition being treated to some extent after administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes, or any other desired change in a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is clinically necessary to provide a significant remission effect of the condition. An effective amount suitable in any individual case can be determined using techniques such as a dose escalation assay.
In this application, the terms "administering", "administration", "administering", and the like refer to a method capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, via the duodenal route, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Administration techniques useful for The compounds and methods described herein are well known to those skilled in The art, for example, in Goodman and Gilman, The pharmaceutical Basis of Therapeutics, current ed.; pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
In the present application, the terms "drug combination", "administering other therapy", "administering other therapeutic agent" and the like refer to a drug treatment obtained by mixing or combining more than one active ingredient, which includes both fixed and unfixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one co-agent to a patient in the form of a single entity or a single dosage form. The term "non-fixed combination" refers to the simultaneous administration, concomitant administration, or sequential administration at variable intervals of at least one compound described herein and at least one synergistic formulation to a patient as separate entities. These also apply to cocktail therapy, for example the administration of three or more active ingredients.
The main advantages of the invention are:
the compound can effectively inhibit the proliferation of KM12-LUC cells.
The compounds of the invention are capable of effectively inhibiting the phosphorylation levels of TRKA, TRKB and TRKC at the cellular level.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Unless otherwise indicated, percentages and parts are percentages and parts by weight.
The test materials and reagents used in the following examples are commercially available without specific reference.
Synthesis of intermediate 1
Figure PCTCN2019111334-APPB-000035
(R, E) -N- (2, 5-difluorobenzylidene) -2-methylpropane-2-sulfinamide
Figure PCTCN2019111334-APPB-000036
2, 5-difluorophenyl (formaldehyde) (5g,35.2mmol) and (R) -2-methylpropane-2-sulfinamide (4.47g,36.9mmol) were dissolved in dichloromethane (50ml), cesium carbonate (8.0g,24.6mmol) was added at room temperature, and then the reaction was warmed to 50 ℃ for 3h, TLC showed completion of the reaction, filtration, the filter cake was washed with dichloromethane, the filtrate was washed with brine, Na2SO 4Drying and spin-drying to obtain (R, E) -N- (2, 5-difluorobenzylidene) -2-methylpropane-2-sulfinamide as a yellow oily liquid (9 g).
(R) -N- ((R) -1- (2, 5-difluorophenyl) -3- (1, 3-dioxan-2-yl) propyl) -2-methylpropane-2-sulfinamide
Figure PCTCN2019111334-APPB-000037
Magnesium turnings (2g,83.3mmol) were dissolved in tetrahydrofuran (72ml) and, under nitrogen, Dibal-H (0.1ml,1.5M,0.15mmol) was added dropwise to the system at 40 ℃ for 0.5H at 40 ℃ and then a solution of 2- (2-bromoethyl) -1, 3-dioxane (14.3g,73.47mmol) in tetrahydrofuran (40ml) was slowly added dropwise to the system with temperature controlled at 40-50 ℃ and after dropping was kept at 40 ℃ for 1H. Heating was removed, the reaction was cooled to-30 ℃ and then (R, E) -N- (2, 5-difluorobenzylidene) -2-methylpropane-2-sulfinamide (9g,36.73mmol) in tetrahydrofuran (40ml) was added dropwise to the system, the temperature was controlled at-30 ℃ to 20 ℃, after completion of the addition, the mixture was stirred at-30 ℃ for 2h, TLC showed completion of the reaction, quenched with 10% aqueous citric acid and controlled at 10 ℃, extracted with dichloromethane, the organic phase was washed with saturated brine, Na2SO 4Drying and spin-drying gave (R) -N- ((R) -1- (2, 5-difluorophenyl) -3- (1, 3-dioxan-2-yl) propyl) -2-methylpropane-2-sulfinamide as a colorless oily liquid (15.8 g).
(R) -2- (2, 5-difluorophenyl) pyrrolidine
Figure PCTCN2019111334-APPB-000038
(R) -N- ((R) -1- (2, 5-difluorophenyl) -3- (1, 3-dioxan-2-yl) propyl) -2-methylpropane-2-sulfinamide (15.8g,43.76mmol) was added to a mixed solution of trifluoroacetic acid (32ml) and water (8ml) at room temperature, stirred at room temperature for 1h, then trifluoroacetic acid (60ml) was added to the system, triethylsilane (15.2g,131.1mmol) was added dropwise to the system, the reaction was allowed to stand overnight at room temperature, LCMS monitored for completion of the reaction, most of the trifluoroacetic acid was spun off, the residue was dissolved in hydrochloric acid (1N,100ml) and stirred for 0.5h, extracted with methyl tert-butyl ether, the organic phase was washed with hydrochloric acid (1N,50ml), the aqueous phases were combined, adjusted to pH 11 with 40% aqueous sodium hydroxide solution and then extracted with dichloromethane, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate and spin-dried to give (R) -2- (2, 5-difluorophenyl) pyrrolidine as an oily liquid (6.7 g).
Pyrazolo [1,5-a ] pyrimidin-5 (4H) -ones
Figure PCTCN2019111334-APPB-000039
Dissolving 1H-pyrazol-5-amine (20 g, 0.24 mol) and 1, 3-dimethylpyrimidine-2, 4(1H,3H) -diketone (35 g, 0.25 mol) in ethanol (200 ml), reducing the temperature of sodium ethoxide (34 g, 0.50 mol) at room temperature, adding the mixture into a reaction system in batches, heating the mixture to 70 ℃ for reaction for 3 hours, monitoring the reaction by LCMS (liquid crystal display system), cooling the reaction system to room temperature, filtering, washing a filter cake by ethanol, washing the filter cake by n-hexane, and drying to obtain pyrazolo [1,5-a ] pyrimidin-5 (4H) -one and a yellow solid (36g, yield: 94%).
3-nitropyrazolo [1,5-a ] pyrimidin-5 (4H) -ones
Figure PCTCN2019111334-APPB-000040
Pyrazolo [1,5-a ] pyrimidin-5 (4H) -one (30 g, 191.1 mol) was added in portions to nitric acid (68%) (150 ml) at room temperature, heated to 80 ℃ for reaction for 3 hours, and LCMS monitored for completion of the reaction, after the reaction system was cooled to room temperature, filtered, the filter cake was washed with water, washed with methanol, and dried to give 3-nitropyrazolo [1,5-a ] pyrimidin-5 (4H) -one as a red solid (28g, yield: 82%).
5-chloro-3-nitropyrazolo [1,5-a ] pyrimidines
Figure PCTCN2019111334-APPB-000041
Reacting 3-nitropyrazolo [1,5-a ]]Pyrimidin-5 (4H) -one (15g, 83.3mmol) and 2, 6-lutidine (17.8 g, 166.7 mmol) were dissolved in acetonitrile (150 mL) and POCl was added at room temperature3(51 g, 333.2 mmol) is added into the system dropwise, then the temperature is raised to 80 ℃ and the mixture is stirred overnight, LCMS monitors that the reaction is finished, the solvent is spun off, the rest is dissolved in dichloromethane (200 ml), the organic phase is washed by water, saturated brine is washed, anhydrous sodium sulfate is dried, and the 5-chloro-3-nitropyrazolo [1,5-a ] is obtained by spin-drying]Pyrimidine, grey solid (15g, yield: 91%).
(R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3-nitropyrazolo [1,5-a ] pyrimidine
Figure PCTCN2019111334-APPB-000042
5-chloro-3-nitropyrazolo [1,5-a ] pyrimidine (2.38 g, 12.02 mmol) and (R) -2- (2, 5-difluorophenyl) pyrrolidine (4 g, 12.62 mmol) were dissolved in ethanol (40ml) and tetrahydrofuran (10 ml), triethylamine (4.2 g, 42 mmol) was added at room temperature, the reaction was heated to 50 ℃ for 2 hours, the reaction was monitored by LCMS, the solvent was removed by spinning off, the residue was dissolved in dichloromethane, the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and spin-dried to give (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3-nitropyrazolo [1,5-a ] pyrimidine as a yellow solid (3.7g, yield: 90%).
(R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1,5-a ] pyrimidin-3-amine
Figure PCTCN2019111334-APPB-000043
Dissolving (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) -3-nitropyrazolo [1,5-a ] pyrimidine (3.7g, 10.72 mmol) in a mixed solution of dichloromethane (50ml), methanol (50ml) and saturated ammonium chloride solution (50ml), adding zinc powder (6.8 g, 107.2 mmol) at room temperature, reacting overnight at room temperature, monitoring the completion of the reaction by LCMS, filtering, washing the filtrate with saturated brine, drying with anhydrous sodium sulfate, spin-drying, passing through a column (dichloromethane/methanol ═ 150/1-50/1) to obtain (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1,5-a ] pyrimidin-3-amine, yellow solid (3.2g, yield: 94%)
1H NMR(400MHz,CDCl3):δ(ppm)8.06(d,J=7.6Hz,1H),7.62(s,1H),7.06-7.00(m,1H),6.91-6.87(m,1H),6.77-6.73(m,1H),5.88(d,J=1.6Hz,1H),5.32-5.29(m,1H),3.92(d,J=7.2Hz,1H),3.75(d,J=7.2Hz,1H),2.78(s,2H),2.48-2.44(m,1H),2.08-2.01(m,3H).
19F NMR(400MHz,CDCl3):δ(ppm)-124.30,-124.35.
Synthesis of intermediate 2
Figure PCTCN2019111334-APPB-000044
1) Synthesis of Compounds 2-3
Figure PCTCN2019111334-APPB-000045
In a single neck flask (100mL), compound 1-1(0.58g, 2.0mmol) and compound 1-2(0.70g, 4.0mmol) were dissolved in EtOH (10.0mL) followed by the addition of AcOH (0.24g, 4.0 mmol). The reaction was stirred at room temperature for 3 hours. Then, add NaBH3CN (0.50g, 8.0 mmol). The resulting reaction solution was stirred at 75 ℃ for 17 hours. Under reduced pressure willThe reaction solution was concentrated to dryness, and the resulting residue was dissolved in 30mL of methylene chloride and 30mL of a saturated ammonium chloride solution. After separation of the organic phase, the aqueous phase is extracted twice with dichloromethane. The combined organic phases were concentrated to dryness and the residue was chromatographed on petroleum ether and ethyl acetate to give 1-3 as a colorless oil (0.72g, 80% yield). LC-MS (ESI +) M/z [ M + H ]]450.32
2) Synthesis of Compounds 2 to 4
Figure PCTCN2019111334-APPB-000046
In a single-necked flask (50mL), compound 1-3(0.72g, 1.6mmol), CH was added2Cl 2(8.0mL) and TFA (1.1g, 9.6 mmol). The reaction was stirred at room temperature for 17 hours. The reaction was concentrated to dryness and the resulting crude product 1-4(1.2g, over 100% yield) was used directly in the next reaction. LC-MS (ESI +) M/z [ M + H ]]350.20
3) Synthesis of Compounds 2 to 5
Figure PCTCN2019111334-APPB-000047
In a single neck flask (100mL) were added compounds 1-4(0.49g, 1.4mmol), MeOH (28mL) and NaOMe in methanol (5.4 mol/L,0.39mL, 2.1 mmol). The reaction solution was stirred at 80 ℃ for 3 hours. The reaction solution was concentrated to dryness, and the residue was dissolved in 20mL of methylene chloride and 20mL of water. After separation of the organic phase, the aqueous phase is extracted twice with dichloromethane. The combined organic phases were concentrated to dryness to give crude 1-5(0.37g, 87% yield). LC-MS (ESI +) M/z [ M + Na ]326.14
4) Synthesis of Compound 2
Figure PCTCN2019111334-APPB-000048
In a single-neck flask (50mL), compound 1-5(0.37g, 1.24mmol), Boc was added2O(0.35g,1.61mmol),Et 3N (0.13g, 1.24mmol) and CH2Cl 2(2.5 mL). The reaction solution was stirred at room temperature for 12 hours. The reaction was concentrated to dryness and the resulting residue was subjected to column chromatography using petroleum ether and ethyl acetate to give 1-6(0.33g, 66% yield) as a yellow oil. LC-MS (ESI +) M/z [ M + Na]426.36
Synthesis of intermediate Compound 3
Figure PCTCN2019111334-APPB-000049
1) Synthesis of Compound 3-2
Figure PCTCN2019111334-APPB-000050
Compound 1(630mg, 2.69mmol), compound 2(698mg, 4.03mmol) and methanol (15mL) were added to a single-necked flask (100mL) and reacted at room temperature for 1 hour, and sodium cyanoborohydride (253mg, 4.03mmol) was added and reacted at room temperature for 16 hours. The reaction was spin dried and the column was washed with dichloromethane and methanol to give 3 as a yellow oil (960mg, 91% yield). LC-MS (ESI) M/z (M + H)+392.31。
2) Synthesis of Compound 3-3
Figure PCTCN2019111334-APPB-000051
Compound 3(960mg, 2.45mmol), dioxane hydrochloride (15mL,60mmol) was added to a single-neck flask (100mL) and the reaction was allowed to react at room temperature for 2 hours. The reaction mixture was spin-dried to give 4 as a yellow oil (980mg, 100% yield). LC-MS (ESI) M/z (M + H)+292.24.
3) Synthesis of Compound 3
Figure PCTCN2019111334-APPB-000052
Compound 4(980mg, 2.45mmol), potassium carbonate (1.014g, 7.35mmol), and methanol (15mL) were added to a single-neck flask (100mL) and the reaction was refluxed at 75 ℃ for 3 hours. The reaction was cooled to room temperature, filtered, the filtrate was spin-dried, and the residue was purified by column chromatography using dichloromethane and methanol to give 5(390mg, yield 61%) as a yellow oil. LC-MS (ESI) M/z (M + H)+260.24。1H NMR(400MHz,DMSO)δ7.26(m,5H),3.59(m,2H),3.22(m,2H),2.80(m,5H),2.65(dd,J=24.8,9.1Hz,2H),2.40(dd,J=15.9,7.9Hz,1H),2.18(ddd,J=12.4,7.8,4.6Hz,1H),1.67(dt,J=12.4,7.3Hz,1H).
EXAMPLE 1N- (5- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1, 7-diazaspiro [4.4] nonane-7-carboxamide
Figure PCTCN2019111334-APPB-000053
To a solution of (R) -5- (2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1,5-a ] pyrimidin-3-amine (40 mg, 0.126 mmol) in dichloromethane (4 ml) was added CDI (41 mg, 0.253 mmol), stirred at room temperature for 2 hours, followed by addition of tert-butyl 1, 7-diazaspiro [4.4] nonane-1-carboxylate (143 mg, 0.632 mmol) and N, N-diisopropylethylamine (163 mg, 1.265 mmol) in dichloromethane (2ml), the reaction was stirred for 2 hours, monitoring of liquid quality showed the formation of the product, concentrated, a hydrochloric acid/1-4 dioxane solution (4 ml, 4N) was added, stirred overnight, concentrated, and the residue was purified by preparative hplc to give the desired product (27.7 mg, 62%) as a yellow solid.
MS-ESI:m/z 468[M+H]+.
1H NMR(400MHz,CD 3OD)δ(ppm)8.24(s,1H),7.87(s,1H),7.16-7.07(m,1H),7.02-6.93(m,1H),6.84-6.80(m,1H),6.40-6.10(m,1H),5.41(s,1H),4.59-4.57(m,1H),4.01-3.91(m,1H),3.80-3.66(m,1H),3.62-3.50(m,2H),3.45-3.32(m,2H),3.05–2.82(m,2H),2.55-2.40(m,1H),2.12-1.96(m,4H),1.94–1.72(m,4H).
Example 27-amino-N- (5- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -2-azaspiro [4.5] decane-2-carboxamide
Figure PCTCN2019111334-APPB-000054
N- (5- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -7-carbonyl-2-azaspiro [4.5] decane-2-carboxamide (30 mg, 0.06 mmol), ammonium acetate (23 mg, 0.30 mmol) were dissolved in methanol (4 ml), stirred at room temperature for 2 hours, sodium cyanoborohydride (11 mg, 0.18 mmol) was added, stirred at room temperature for 2 hours, concentrated, and the residue was purified by preparative high performance liquid chromatography to give the desired product (27.7 mg, 62%) as a yellow solid.
MS-ESI:m/z 496[M+H]+.
1H NMR(400MHz,CD 3OD)δ(ppm)8.25(d,J=4.0Hz,1H),7.87(s,1H),7.14-7.08(m,1H),7.01-6.93(m,1H),6.84-6.80(m,1H),6.15(brs,1H),5.41(s,1H),4.00-3.91(m,1H),3.75-3.73(m,1H),3.49-3.45(m,2H),3.22-3.15(m,1H),2.88-2.77(m,1H),2.51-2.41(m,1H),2.07-1.11(m,16H).
EXAMPLE 3N- (5- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -1, 4-dimethyl-5-carbonyloctahydropyrrolo [3,4-e ] [1,4] diazepine-7 (1H) -carboxamide
Figure PCTCN2019111334-APPB-000055
N- (5- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) pyrazolo [1,5-a ] pyrimidin-3-yl) -4-methyl-5-carbonyloctahydropyrrolo [3,4-e ] [1,4] diazepin-7 (1H) -carboxamide (14 mg, 0.027 mmol), aqueous formaldehyde (1 ml) and acetic acid (2 drops) were dissolved in methanol (3 ml), stirred at room temperature for 2 hours, sodium cyanoborohydride (5 mg, 0.082 mmol) was added, stirred at room temperature for 1 hour, concentrated, and the residue was purified by preparative high performance liquid chromatography to give the desired product (27.7 mg, 62%) as a yellow solid.
MS-ESI:m/z 525[M+H]+.
1H NMR(400MHz,CD 3OD)δ(ppm)8.25(d,J=6.4Hz,1H),7.87(d,J=4.8Hz, 1H),7.16-7.10(m,1H),6.99-6.93(m,1H),6.84-6.79(m,1H),6.14(brs,1H),5.39(brs,1H),4.00-3.87(m,4H),3.80-3.71(m,2H),3.45-3.33(m,2H),3.01(s,3H),3.01-2.95(m,2H),2.67-2.59(m,1H),2.49-2.36(m,2H),2.32(s,3H),2.07-1.96(m,3H).
The corresponding starting materials were replaced by analogous conditions to those of examples 1 to 3, giving the compounds shown in the following table:
Figure PCTCN2019111334-APPB-000056
Figure PCTCN2019111334-APPB-000057
comparative example 12- (1- (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazine-3-carbonyl) -3-hydroxypyrrolidin-3-yl) -N-methylacetamide
Figure PCTCN2019111334-APPB-000058
Preparation of the title Compound under analogous conditions to example 1
MS(ESI):m/z=485[M+H]+.
1H NMR(400MHz,CD3OD)δ(ppm)7.75-7.70(m,1H),7.65(t,J=5.5Hz,1H),7.18–7.04(m,1H),7.04–6.78(m,3H),5.38-5.32(m,1H),4.08–3.43(m,6H),2.78–2.60(m,3H),2.58-2.40(m,2H),2.35-2.15(m,1H),2.14–1.69(m,5H).
Comparative example 2 (6- ((R) -2- (2, 5-difluorophenyl) pyrrolidin-1-yl) imidazo [1,2-b ] pyridazin-3-yl) (3- ((dimethylamino) methyl) -4-hydroxypyrrolidin-1-yl) methanone
Figure PCTCN2019111334-APPB-000059
Preparation of the title Compound under analogous conditions to example 1
MS(ESI):m/z=471[M+H]+.
1H NMR(400MHz,CD3OD)δ(ppm)7.77-7.70(m,1H),7.67-7.65(m,1H),7.18–7.08(m,1H),7.00-6.80(m,2H),5.34(t,J=9.4Hz,1H),4.56(s,1H),4.15-4.05(m,1H),4.00-3.63(m,4H),3.60-3.45(m,1H),3.44–3.33(m,1H),3.15-2.88(m,1H),2.56–2.40(m,2H),2.39–1.92(m,9H).
Biological example 1: TRKA, TRKB, TRKC kinase in vitro activity assay
Experimental Material
Recombinant human TRKA, TRKB, TRKC proteins were purchased from Carna Biosciences. HTRF kinEASE TK kit was purchased from CisbioBioassays. The BioTek microplate reader Synergy Neo 2 plate was used.
Experimental methods
Test compounds were diluted in 3-fold concentration gradient to a final concentration of 1 μ M to 0.05nM 10 concentrations, two replicate wells per concentration; the content of DMSO in the assay reaction was 1%.
TRKA enzyme reaction:
0.2 ng/. mu.l TRKA protein kinase, 1. mu.M TK Substrate-biotin (Substrate-biotin) polypeptide Substrate, 14.68. mu.M ATP, 1 Xenzyme buffer, 5mM MgCl21mM DTT. The detection plate is White Proxiplate384-Plus plate (PerkinElmer), the reaction is carried out for 40 minutes at room temperature, and the reaction system is 10 mu l.
TRKB enzyme reaction:
0.037 ng/. mu.l TRKB protein kinase, 1. mu.M TK Substrate-biotin (Substrate-biotin) polypeptide Substrate, 4.77. mu.M ATP, 1 Xenzyme buffer, 5mM MgCl2,1mMMnCl 21mM DTT. The assay plate was a White Proxiplate384-Plus plate (PerkinElmer) and reacted at room temperature for 50 minutes in a 10. mu.l reaction system.
TRKC enzyme reaction:
0.037ng/μl TRKprotein C kinase, 1. mu.M TK Substrate-biotin (Substrate-biotin) polypeptide Substrate, 25.64. mu.M ATP, 1 Xenzyme buffer, 5mM MgCl21mM DTT. The detection plate is White Proxiplate384-Plus plate (PerkinElmer), the reaction is carried out for 40 minutes at room temperature, and the reaction system is 10 mu l.
Reaction detection:
mu.l of detection reagent were added to the reaction plate, containing SA-XL665 at a final concentration of 0.125. mu.M and 1 XTK-antibody 5. mu.l, incubated overnight at room temperature, Synergy Neo 2 read plate.
Data analysis
The 665/620Ratio value was converted into an inhibition Ratio (%) (1-Ratio) by the following equationtest/Ratio max)×100%。Ratio maxRatio as a positive control without test CompoundtestThe values were measured for each concentration of different compounds. IC50(nM) data were obtained by 4-parameter curve fitting, as shown in Table 1.
TABLE 1
Compound (I) TRKA(nm) TRKB(nm) TRKC(nm)
Example 1 ++ + ++
Example 2 +++ +++ +++
Example 3 +++ +++ +++
Example 4 ++ ++ ++
Example 5 +++ ++ +++
Example 6 +++ ++ +++
Example 7 ++ + ++
Example 8 ++ ++ ++
Example 9 +++ +++ +++
Comparative example 1 + - -
Comparative example 2 - - -
Wherein, + + + + denotes IC50< 10nm, + denotes 10nm < IC50< 100nm, + denotes 100nm < IC50< 500nm, -represents IC50>500nm
Bioassay example 2 detection of cytological level of TRK kinase Activity by the Elisa method
NIH-3T3 cell line stably expressing normal TRKA or TRKB or TRKC was constructed by plasmid transfection.
Cells were seeded on the first day in 96-well cell culture plates at 10000 cells/well in normal medium (DMEM + 10% FBS). The next day, the medium containing 0.5% FBS was starved overnight. On the third day, cells were treated with different concentrations of test compound for 1 hour and then stimulated with 100ng/ul growth factor for 10 minutes (NGF for TRKA, BDNF for TRKB, NT-3 for TRKC). Placing the cell culture plate on ice; the supernatant was removed and rinsed once with pre-cooled PBS. The lysis buffer (cellsingailing Technology) attached to the elisa kit was diluted with double distilled water and protease and phosphatase inhibitors were added. The prepared cell lysate was added to the well plate and allowed to stand on ice for 20 minutes. The cell lysate was blown several times by the rifle and transferred to antibody pre-coated strips, capped and incubated 4 degrees overnight. The remaining steps are performed with reference to the methods provided in the elisa kit (e.g. as described in Cellsignaling Technology # 7212C).
The results show that the compound of the invention can effectively inhibit TRKA and TRKB phosphorylation levels at a cytological level, and particularly shown in a table 2.
TABLE 2
Compound (I) TRKA(nm) TRKB(nm)
Example 2 ++ ++
Bioassay example 3 KM12-LUC cell proliferation assay
A human colon cancer cell line KM12-LUC (LUC, stably expressing Luciferace) containing a TPM3-NTRK1 fusion gene is used as a model for evaluating the cytological pharmacodynamics of a compound to be tested. The TRK fusion gene in KM12-LUC cells makes the cells independent of stimulation of extracellular growth factors, and can continuously and spontaneously activate downstream signaling pathways MAPK-ERK, PI3K-AKT and other signaling pathways closely related to cell proliferation. Therefore, inhibition of TRK activity in KM12-LUC cells significantly inhibited cell proliferation.
The method comprises the following steps: the first day, cells were seeded in 384-well plates, 2000 cells/well; adding the compounds to be detected with different concentrations on the next day; on the fifth day, cell activity was measured by adding CellTiter-glo (Promega), and the cell proliferation inhibition rate was calculated for 72 hours. Statistical analysis with prism5 and IC of test compound50The value is obtained.
The results show that the compound of the invention can effectively inhibit the proliferation of KM12-LUC cells, and the details are shown in Table 3
TABLE 3
Compound (I) KM12-LUC(nm)
Example 2 ++
Bioassay example 4 mutant TRKA (G595R), TRKA (G667C) and TRKC (G623R) kinase in vitro Activity assay
Experimental Material
Recombinant human TRKA (G595R), TRKA (G667C), and TRKC (G623R) proteins were purchased from SignalChem. HTRF kinEASE TKkit was purchased from CisbioBioassays. The BioTek microplate reader Synergy Neo 2 plate was used.
Experimental methods
Test compounds were diluted in 4-fold concentration gradient to a final concentration of 10 concentrations from 1 μ M to 0.004nM, two duplicate wells per concentration; the content of DMSO in the assay reaction was 1%.
TRKA (G595R) enzyme reaction:
0.12 ng/. mu.l TRKA (G595R) protein kinase, 1. mu.M TK Substrate-biotin polypeptide Substrate, 4.5. mu.M ATP, 1 × enzymic buffer, 5mM MgCl21mM DTT. The detection plate is White Proxiplate384-Plus plate (PerkinElmer), the reaction is carried out for 30 minutes at room temperature, and the reaction system is 10 mu l.
TRKA (G667C) enzyme reaction:
0.026 ng/. mu.l TRKA (G667C) protein kinase, 1. mu.M TK Substrate-biotin polypeptide Substrate, 5.5. mu.M ATP, 1 × enzymic buffer, 5mM MgCl21mM DTT. The detection plate is White Proxiplate384-Plus plate (PerkinElmer), the reaction is carried out for 30 minutes at room temperature, and the reaction system is 10 mu l.
TRKC (G623R) enzymatic reaction:
1.0 ng/. mu.l TRKC (G623R) protein kinase, 1. mu.M TK Substrate-biotin polypeptide Substrate, 62.9. mu.M ATP, 1 × enzymic buffer, 5mM MgCl21mM DTT. The assay plate was a White Proxiplate384-Plus plate (PerkinElmer) and reacted at room temperature for 50 minutes in a 10. mu.l reaction system.
Reaction detection:
add 10. mu.l of assay reagent to the reaction plate, containing SA-XL665 at a final concentration of 0.125. mu.M and 1 XTK-Antibody 5. mu.l, incubated overnight at room temperature, Synergy Neo 2 read plate.
Data analysis
The value of 665/620Ratio minus the value of the enzyme-free negative control well was converted to the percent inhibition (%) (1-Ratio) by the following equationtest/Ratio max)×100%。Ratio maxRatio as a positive control without test CompoundtestThe values were measured for each concentration of different compounds. IC50(nM) data were obtained by 4-parameter curve fitting, as shown in Table 4.
TABLE 4
Compound (I) TRKA(G595R)(nm)
Example 1 ++
Example 2 ++
Example 3 ++
Example 4 ++
Example 5 +
Example 6 ++
Example 7 ++
Example 8 +
Example 9 ++
Comparative example 1 ++
Comparative example 2 ++
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (10)

  1. A compound of formula I or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2019111334-APPB-100001
    wherein the content of the first and second substances,
    ring a has a structure represented by the following formula:
    Figure PCTCN2019111334-APPB-100002
    each X4Each independently is C or N;
    each X5Each independently is CR7Or N; wherein R is7Is hydrogen, deuterium, halogen, NH2Or OH;
    R 1、R 2taken together with the nitrogen atom to which they are attached to form a substituted or unsubstituted 6-12 membered fused ring (including bicyclic fused ring, tricyclic fused ring), bridged ring or spiro ring; wherein said fused, bridged or spiro ring contains 1 to 3 heteroatoms selected from nitrogen atoms, oxygen atoms and sulfur atoms;
    R Ais composed of
    Figure PCTCN2019111334-APPB-100003
    Wherein the content of the first and second substances,
    X 1、X 2、X 3each independently is CR6Or N; r6Is hydrogen, deuterium, hydroxy, halogen, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy;
    n is 1,2 or 3;
    each R is3Each independently is hydrogen, deuterium, hydroxy, halogen, trifluoromethyl, cyano, amino, amido, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy;
    or n is 2 and two R3co-form-O- (CH)2) m-O-; m is an integer of 1 to 4;
    R Bis a substituted or unsubstituted 4-7 membered heterocyclic ring (said heterocyclic ring containing 1 to 3 hetero atoms selected from the group consisting of a nitrogen atom, an oxygen atom and a sulfur atom); - (CH)2) m1-、-O-(CH 2) m2-O-; wherein m1 and m2 are each independently an integer of 1 to 4;
    or RBIs-substituted or not substitutedsubstituted-C1-C10 alkylene-L2- (wherein, L)2Is NH, O, S or S (O)2) Substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy;
    unless otherwise specified, "substituted" means substituted with one or more (e.g., 2, 3,4, etc.) substituents selected from the group consisting of: deuterium, halogen, C1-C6 alkoxy, halogenated C1-C6 alkyl, halogenated C1-C6 alkoxy, halogenated C3-C8 cycloalkyl, methylsulfonyl, -S (═ O)2NH 2Oxo (═ O), -CN, hydroxy, -NH2Carboxy, C1-C6 amido (-C (═ O) -N (Rc)2or-NH-C (═ O) (Rc), Rc being H or C1-C5 alkyl), C1-C6 alkyl- (C1-C6 amido),
    Figure PCTCN2019111334-APPB-100004
    Or a substituted or unsubstituted group selected from: C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 amino, C6-C10 aryl, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O, 5-12 membered heterocyclyl having 1-3 heteroatoms selected from N, S and O, - (CH) 6 alkyl2) -C6-C10 aryl, - (CH)2) - (5-to 10-membered heteroaryl having 1 to 3 heteroatoms selected from N, S and O), and the substituents are selected from the group consisting of: halogen, C1-C6 alkyl, C1-C6 alkoxy, oxo, -CN, -NH2-OH, C6-C10 aryl, C1-C6 amine, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O;
    the dotted line represents a chemical bond or nothing.
  2. A compound according to claim 1, or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof: characterized in that ring A is a bicyclic 9-10 membered heteroaromatic ring; wherein the heteroaromatic ring contains 1-3 nitrogen atoms.
  3. A compound according to claim 1, or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof: its special featureCharacterized in that R is1、R 2Together with the nitrogen atom to which they are attached form a substituted or unsubstituted 6-12 membered bicyclic, bridged or spirocyclic ring; the fused ring, bridged ring or spiro ring contains 1-3 nitrogen atoms.
  4. A compound according to claim 1, or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof: characterized in that R isASelected from the group consisting of:
    Figure PCTCN2019111334-APPB-100005
    in each formula, X1、X 2The definition is the same as the previous definition;
    on the aromatic ring, n is 1,2 or 3; and R is3Is hydrogen, deuterium, hydroxy, halogen, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy; or n is 2 and two R3co-form-O- (CH)2) m-O-; m is an integer of 1 to 4;
    r in other positions3、R 4、R 5Each independently hydrogen, deuterium, hydroxy, halogen, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy.
  5. A compound according to claim 1, or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof: characterized in that R isAIs composed of
    Figure PCTCN2019111334-APPB-100006
    Wherein, X1Is CR6Or N; x2、X 3Each independently is CR6
    n is 1,2 or 3; and R is3Is hydrogen, deuterium, hydroxy, halogen, substituted or unsubstituted C1-C6 alkyl, or substituted or unsubstituted C1-C6 alkoxy; (ii) a Or n is 2 and two R3co-form-O- (CH)2) m-O-; m is an integer of 1 to 4.
  6. A compound according to claim 1, or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof: characterized in that it is selected from the following group:
    Figure PCTCN2019111334-APPB-100007
    Figure PCTCN2019111334-APPB-100008
  7. a pharmaceutical composition comprising a therapeutically effective amount of a compound of claim 1, or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  8. Use of the compound of claim 1 or a stereoisomer or racemate thereof or a pharmaceutically acceptable salt thereof or the pharmaceutical composition of claim 7, for the preparation of a medicament for the prevention and treatment of a disease associated with abnormal TRK function.
  9. A TRK inhibitor, wherein said inhibitor comprises a compound of claim 1, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof.
  10. A method for preparing a compound of claim 1, or a stereoisomer or racemate thereof, or a pharmaceutically acceptable salt thereof, comprising the steps of:
    Figure PCTCN2019111334-APPB-100009
    reacting a compound of formula Ia with a compound of formula Ib in an inert solvent, thereby forming a compound of formula I;
    wherein R is1Is C1-C6 alkyl; the remaining groups are as defined in claim 1.
CN201980067895.1A 2018-10-15 2019-10-15 Preparation and application of protein receptor kinase inhibitor Pending CN113242858A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201811201489.3A CN111039947A (en) 2018-10-15 2018-10-15 Preparation and application of protein receptor kinase inhibitor
CN2018112014893 2018-10-15
PCT/CN2019/111334 WO2020078360A1 (en) 2018-10-15 2019-10-15 Preparation and uses of protein receptor kinase inhibitor

Publications (1)

Publication Number Publication Date
CN113242858A true CN113242858A (en) 2021-08-10

Family

ID=70230441

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201811201489.3A Pending CN111039947A (en) 2018-10-15 2018-10-15 Preparation and application of protein receptor kinase inhibitor
CN201980067895.1A Pending CN113242858A (en) 2018-10-15 2019-10-15 Preparation and application of protein receptor kinase inhibitor

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN201811201489.3A Pending CN111039947A (en) 2018-10-15 2018-10-15 Preparation and application of protein receptor kinase inhibitor

Country Status (2)

Country Link
CN (2) CN111039947A (en)
WO (1) WO2020078360A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116003469B (en) * 2022-03-23 2023-10-17 南京知和医药科技有限公司 Preparation and use methods of pyrimidinyl antiviral compounds
CN117343064B (en) * 2022-07-05 2024-04-16 南京知和医药科技有限公司 Preparation and application of pyrimidine derivative with antiviral effect

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102224153A (en) * 2008-09-22 2011-10-19 阵列生物制药公司 Substituted imidazo[1,2b]pyridazine compounds as trk kinase inhibitors
CN102264736A (en) * 2008-10-22 2011-11-30 阵列生物制药公司 Substituted pyrazolo[1,5-a]pyrimidine compounds as trk kinase inhibitors
WO2019174598A1 (en) * 2018-03-14 2019-09-19 Fochon Pharmaceuticals, Ltd. SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102224153A (en) * 2008-09-22 2011-10-19 阵列生物制药公司 Substituted imidazo[1,2b]pyridazine compounds as trk kinase inhibitors
CN102264736A (en) * 2008-10-22 2011-11-30 阵列生物制药公司 Substituted pyrazolo[1,5-a]pyrimidine compounds as trk kinase inhibitors
WO2019174598A1 (en) * 2018-03-14 2019-09-19 Fochon Pharmaceuticals, Ltd. SUBSTITUTED (2-AZABICYCLO [3.1.0] HEXAN-2-YL) PYRAZOLO [1, 5-a] PYRIMIDINE AND IMIDAZO [1, 2-b] PYRIDAZINE COMPOUNDS AS TRK KINASES INHIBITORS

Also Published As

Publication number Publication date
CN111039947A (en) 2020-04-21
WO2020078360A1 (en) 2020-04-23

Similar Documents

Publication Publication Date Title
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
EP3712151B1 (en) (s)-1&#39;-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4&#39;-piperidine]-5-amine as shp2 inhibitor for the treatment of cancer
WO2021018287A1 (en) Spiroaromatic compound, preparation and application thereof
KR101686685B1 (en) Pyrazolopyrimidine jak inhibitor compounds and methods
WO2019158019A1 (en) Pyrimidine-fused cyclic compound, preparation method therefor and application thereof
KR101469334B1 (en) N-9-substituted purine compounds, compositions and methods of use
CN106187915A (en) There is inhibitor of ALK Yu EGFR double activity and its preparation method and application
CN114981268A (en) Pyrimidine-4 (3H) -ketone heterocyclic compound, preparation method and application thereof in medicine and pharmacology
TWI822868B (en) FGFR4 inhibitors, pharmaceutical compositions containing them and uses thereof
WO2020156437A1 (en) Akt inhibitor
CN111518100A (en) Cyclopropenoarylbenzofuran substituted nitrogen heteroaryl compound and application thereof
CN113242858A (en) Preparation and application of protein receptor kinase inhibitor
AU2010229140A1 (en) Novel aminopyridine derivatives having Aurora A selective inhibitory action
CN112300173B (en) Nitrogen-containing polycyclic compounds, preparation method and application
CN115477640A (en) Pyridazinones as PARP7 inhibitors
CN111747954B (en) Pyrazine compounds and uses thereof
CN114641476B (en) Preparation and application of protein receptor kinase inhibitors
JP2022501397A (en) PD-L1 antagonist compound
WO2023143147A1 (en) Pyridazopyridone compounds, pharmaceutical composition thereof and use thereof
CN112279837A (en) Pyrazine compounds and uses thereof
WO2019057112A1 (en) 2-substituted pyrazole amino-4-substituted amino-5-pyrimidine formamide compound, composition, and application thereof
CN111763217B (en) Thieno-nitrogen heterocyclic compounds, preparation method and application
WO2023046030A1 (en) Egfr small-molecule inhibitor, pharmaceutical composition containing same, and use thereof
WO2023116763A1 (en) Pyridazine compound, and pharmaceutical composition and use thereof
WO2024088296A1 (en) Piperidinopyrimidine derivative, preparation method therefor and use thereof in medicine

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20210810

WD01 Invention patent application deemed withdrawn after publication